• Aucun résultat trouvé

Effects of endosulfan on hepatoma cell adhesion: Epithelial-mesenchymal transition and anoikis resistance

N/A
N/A
Protected

Academic year: 2021

Partager "Effects of endosulfan on hepatoma cell adhesion: Epithelial-mesenchymal transition and anoikis resistance"

Copied!
13
0
0

Texte intégral

(1)

HAL Id: hal-01137000

https://hal.archives-ouvertes.fr/hal-01137000

Submitted on 30 Mar 2015

HAL is a multi-disciplinary open access

archive for the deposit and dissemination of

sci-entific research documents, whether they are

pub-lished or not. The documents may come from

teaching and research institutions in France or

abroad, or from public or private research centers.

L’archive ouverte pluridisciplinaire HAL, est

destinée au dépôt et à la diffusion de documents

scientifiques de niveau recherche, publiés ou non,

émanant des établissements d’enseignement et de

recherche français ou étrangers, des laboratoires

publics ou privés.

Effects of endosulfan on hepatoma cell adhesion:

Epithelial-mesenchymal transition and anoikis resistance

Ludovic Peyre, Nathalie Zucchini-Pascal, Georges de Sousa, Roger Rahmani

To cite this version:

Ludovic Peyre, Nathalie Zucchini-Pascal, Georges de Sousa, Roger Rahmani. Effects of endosulfan

on hepatoma cell adhesion: Epithelial-mesenchymal transition and anoikis resistance. Toxicology,

Elsevier, 2012, 300 (1-2), pp.19-30. �10.1016/j.tox.2012.05.008�. �hal-01137000�

(2)

Contents

lists

available

at

SciVerse

ScienceDirect

Toxicology

j o u

r n

a l

h o m

e p a g e :

w w w . e l s e v i e r . c o m / l o c a t e / t o x i c o l

Effects

of

endosulfan

on

hepatoma

cell

adhesion:

Epithelial–mesenchymal

transition

and

anoikis

resistance

Ludovic

Peyre,

Nathalie

Zucchini-Pascal

,

Georges

de

Sousa,

Roger

Rahmani

LaboratoiredeToxicologieCellulaireetMoléculairedesXénobiotiques,INRA,UMR1331TOXALIMSophiaAntipolis,06903SophiaAntipolis,France

a

r

t

i

c

l

e

i

n

f

o

Articlehistory:

Received28March2012

Receivedinrevisedform10May2012 Accepted12May2012

Available online 5 June 2012

Keywords: Endosulfan Anoikis EMT Hepatocarcinoma HepG2

a

b

s

t

r

a

c

t

EndosulfanisanorganochlorinepesticidecommonlyusedinagricultureyetclassifiedbytheStockholm Conventionin2011asapersistentorganicpollutant(POP).Itspotentialtoxicitymakesitscontinueduse amajorpublichealthconcern.Despitestudiesinlaboratoryanimals,themolecularmechanisms under-lyingthecarcinogeniceffectsofendosulfaninhumanliverremainpoorlyunderstood.Inthisstudy,we investigatedthephenotypicaleffectsofendosulfanonHepG2livercells.First,wefoundthatendosulfan disruptedtheanoikisprocess.Indeed,cellsexposedtoendosulfanwereinitiallysensitizedtoanoikisand thereafterrecoveredtheirresistancetothisprocess.Thisphenomenonoccurredinparalleltothe induc-tionoftheepithelialtomesenchymal(EMT)process,asdemonstratedby:(1)reorganizationoftheactin cytoskeletontogetherwithactivationoftheFAKsignalingpathway;(2)repressionofE-cadherin expres-sion;(3)inductionofSnailandSlug;(4)activationoftheWNT/␤-cateninpathway;and(5)inductionand reorganizationofmesenchymalmarkers(S100a4,vimentin,fibronectin,MMP-7).Secondly,despitethe acquisitionofmesenchymalcharacteristics,HepG2cellsexposedtoendosulfanfailedtomigrate.This incapacitytoacquireamotilephenotypecouldbeattributedtoadisruptionoftheinteractionbetween theECMandthecells.Takentogether,theseresultsindicatethatendosulfanprofoundlyaltersthe pheno-typeoflivercellsbyinducingcelldetachmentandpartialEMTaswellasdisruptingtheanoikisprocess. Alltheseeventsaccount,atleastinpart,forthecarcinogenicpotentialofendosulfaninliver.

© 2012 Elsevier Ireland Ltd. All rights reserved.

1.

Introduction

Since

the

1950s,

organochlorine

pesticides

(OCPs)

such

as

the

chlorinated

cyclodiene

endosulfan

have

been

employed

exten-sively

in

agriculture,

viticulture,

horticulture,

domestic

and

public

health.

Endosulfan

is

sold

under

different

trade

names

(Thiodan

®

,

Rocky

®

,

Thionex

®

.

.

.)

and

corresponds

to

a

mixture

of

70%

endosulfan-

and

30%

endosulfan-

␤.

While

it

does

have

beneficial

effects

on

crops,

it

also

represents

a

potential

source

of

environ-mental

contamination

and

risk

to

public

health

(

Silva

and

Gammon,

Abbreviations: Ab,antibody;DAPI,4,6-di-amidino-2-phenylindole;DMSO,

dimethylsulfoxide;ECM,extracellularmatrix;EMT,epithelialtomesenchymal tran-sition;FAK,focaladhesionkinase;FBS,foetalbovineserum;HCC,hepatocellular carcinoma;LEF1/TCF-1,lymphoidenhancerfactor1/Tcellfactor1;MMP, met-alloprotease;MTT,3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyltetrazoliumbromide; Poly-HEMA,poly2-hydroxyethylmethacrylate;POP,persistentorganicpollutant; ROCK,Rho-associatedproteinkinase;WNT,winglessintegrationsite;XIAP,X-linked inhibitorofapoptosisprotein.

∗ Correspondingauthorsat:LaboratoiredeToxicologieCellulaireetMoléculaire desXénobiotiques,INRA,UMR1331,400routedesChappes,BP167,06903Sophia Antipolis,France.Tel.:+33492386548;fax:+33492386401.

E-mailaddresses:zucchini@sophia.inra.fr(N.Zucchini-Pascal), rahmani@sophia.inra.fr(R.Rahmani).

2009

).

Indeed,

endosulfan

is

a

persistent

organic

pollutant

(POP)

that

can

build

up

in

the

environment

and

bioaccumulate

through

the

food

chain.

Its

persistent

and

bioaccumulative

properties

enable

it

to

become

stored

in

fats

where

it

persists

for

a

long

time.

Endo-sulfan

is

readily

absorbed

by

humans

via

the

stomach,

the

skin,

the

lungs,

and

the

placenta

(

Lopez-Espinosa

et

al.,

2008

).

This

OCP

is

one

of

the

most

toxic

pesticides

currently

on

the

market

and

is

responsible

for

a

large

number

of

poisoning-related

deaths

(

Moses

and

Peter,

2010

).

In

animals,

it

is

toxic

to

the

liver,

kidney,

nervous

system,

blood

system,

immune

system

and

reproductive

organs

(

Hashizume

et

al.,

2010;

Karatas

et

al.,

2006;

Ahmed

et

al.,

2011;

Merhi

et

al.,

2010;

Briz

et

al.,

2011;

Chan

et

al.,

2006;

Choudhary

et

al.,

2003;

Aggarwal

et

al.,

2008

).

Due

to

the

risk

of

adverse

effects

on

human

health,

the

World

Health

Organization

(WHO)

classi-fied

endosulfan

as

a

moderately

hazardous

Class

2

pesticide

while

the

Environmental

Protection

Agency

(EPA)

considered

it

as

“highly

acutely

toxic”

(Category

I).

Despite

then

being

banned

in

more

than

62

countries

and

by

the

Stockholm

Convention

in

2011,

it

is

still

used

extensively

(

Weber

et

al.,

2010

).

An

interdiction

for

use

will

be

effective

in

2017.

While

the

potential

carcinogenic

effects

of

endosulfan

remain

a

matter

of

debate,

in

vitro

studies

have

revealed

mechanisms

induced

by

endosulfan

that

are

implicated

in

tumor

development

and

progression

in

testes,

breast

and

liver

(

Hardell

and

Eriksson,

0300-483X/$–seefrontmatter © 2012 Elsevier Ireland Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tox.2012.05.008

(3)

Table1

PrimaryantibodiesusedforWesternblotandimmunofluorescencestaining.

Antigen Phosphorylationsite Source/type Manufacturer DilutionWB DilutionIF

ROCK1 RabbitmAba Epitomics 1:2000 1:200

pFAK Tyr925 RabbitpAbb Cellsignaling 1:2000

FAK RabbitpAb Cellsignaling 1:2000

Vimentin RabbitmAb Epitomics 1:200

TCF1/LEF-1a RabbitmAb Epitomics 1:10,000

Gapdh RabbitmAb Cellsignaling 1:15,000

E-cadherin RabbitmAb Epitomics 1:5000

␤-Catenin RabbitmAb SantaCruz 1:2000 1:400

Fibronectin RabbitmAb Epitomics 1:1000 1:100

Snail RabbitpAb SantaCruz 1:200

Slug RabbitmAb Cellsignaling 1:300

amAb,monoclonalantibody. b pAb,polyclonalantibody.

2003;

Høyer

et

al.,

2002;

Fransson-Steen

et

al.,

1992;

Perez-Carreon

et

al.,

2009

).

An

example

of

such

a

mechanism

is

the

disruption

of

gap

junctions

(

Ruch

et

al.,

1990

).

Such

gap

junctional

intercellu-lar

communication

uncouplers

are

responsible

for

many

cellular

dysfunctions

and

multiple

diseases

including

cancer

(

Ehrlich

et

al.,

2006;

Trosko,

2011

).

In

addition,

the

genotoxic

potential

of

endo-sulfan

has

been

described

in

HepG2

cells

(

Hashizume

et

al.,

2010;

Li

et

al.,

2011

)

and

exposure

to

sub-lethal

doses

of

endosulfan

and

its

metabolites

causes

DNA

damage

and

mutations

(

Antherieu

et

al.,

2007;

Bajpayee

et

al.,

2006

).

One

study

using

a

computa-tional

quantum

chemical

model

indicated

that

endosulfan

and

all

its

metabolites

have

carcinogenic

potential

(

Bedor

et

al.,

2010

).

However,

the

exact

molecular

mechanisms

underlying

the

toxic

and

carcinogenic

effects

of

endosulfan

in

liver

cells

remain

poorly

understood.

Epithelial

to

mesenchymal

transition

(EMT)

is

a

highly

coor-dinated

multistep

process

during

which

epithelial

cells

acquire

mesenchymal

fibroblast-like

properties.

It

has

been

implicated

in

a

variety

of

diseases

including

fibrosis

and

in

the

progression

of

carcinoma

(

Jou

and

Diehl,

2010

).

This

fundamental

program

also

plays

a

key

role

in

the

critical

phases

of

embryonic

development

and

in

adults

it

contributes

on

the

one

hand

to

physiological

pro-cesses

such

as

tissue

repair

and

on

the

other

pathological

conditions

(

Kalluri

and

Weinberg,

2009

).

The

morphological

changes

imposed

are

governed

by

multiple

molecular

mechanisms

including

the

loss

of

epithelial

proteins

such

as

E-cadherin

(

Cavallaro

and

Christofori,

2004;

Huber

et

al.,

2005

)

that

leads

to

the

reduction

in

cell-to-cell

junctions

(adherens

and

gap

junctions),

the

loss

of

cell

polarity,

and

the

gain

of

mesenchymal

markers

such

as

the

extracellular

matrix

compound

fibronectin,

the

intermediate

filament

protein

vimentin

and

S100a4

(

Peinado

et

al.,

2004;

Kim

et

al.,

2011;

Ogunwobi

and

Liu,

2011

).

All

these

steps

lead

to

the

acquisition

of

motile

and

invasive

properties

(

Matsuo

et

al.,

2009

).

E-cadherin

is

thought

to

be

downregulated

via

several

repres-sors

acting

either

indirectly

(e.g.

Twist,

Goosecoid)

or

directly

by

binding

to

and

repressing

the

E-cadherin

promoter

(e.g.

Snail,

Slug,

Zeb)

(

Peinado

et

al.,

2007;

Yang

and

Weinberg,

2008

).

E-cadherin

repression

is

frequently

accompanied

by

the

activation

of

the

␤-catenin/Wnt

signaling

cascade

(

Fransvea

et

al.,

2008;

van

Zijl

et

al.,

2009a,b

).

Indeed,

during

EMT,

the

cytoplasmic

stabilization

of

␤-catenin

occurring

via

WNT

activation

signaling

and

GSK3-

repression

(

Medici

et

al.,

2008

),

leads

to

the

formation

of

a

nuclear

complex

TCF/LEF/

␤-catenin

that

modulates

the

expression

of

tar-get

genes

such

as

fibronectin,

MMP7

or

S100a4

(

Iwai

et

al.,

2010

).

Communications

between

cells

and

the

new

extracellular

matrix

environment

are

mainly

mediated

by

the

activation

of

the

focal

adhesion

kinase-FAK-transduction

pathway

(

Chatzizacharias

et

al.,

2008;

Cicchini

et

al.,

2008

).

The

liver

is

the

most

sensitive

tissue

to

xenobiotic

exposure

and

constitutes

the

main

target

of

endosulfan

toxicity.

Thus,

we

investigated

the

cellular

and

molecular

effects

of

endosulfan

in

human

liver

cells

using

the

HepG2

cell

line.

These

cells

retain

many

cellular

functions

allowing

the

study

of

several

molecular

pro-cesses

among

which

cell

plasticity

and

anoikis

(

Roe

et

al.,

1993

).

In

this

study,

we

show

that

endosulfan

permits

a

transient

anoikis

induction

that

is

followed

by

recovery

of

anoikis

resistance.

This

process

occurs

in

parallel

to

partial

EMT

and

is

characterized

by

the

repression

of

E-cadherin

expression,

the

loss

of

adherent

junctions,

cytoskeleton

reorganization,

extra-cellular

matrix

(ECM)

reshuffle,

WNT/

␤-catenin

pathway

activation

and

the

gain

of

mesenchymal

markers

(vimentin,

fibronectin,

S100a4).

Despite

evidence

of

all

these

events,

the

cells

remain

unable

to

acquire

a

motile

phenotype.

Thus

the

present

study

shows

that

endosulfan

transiently

sensitizes

HepG2

cells

to

anoikis

before

allowing

a

later

recovery

of

resistance

to

this

process

that

coincides

with

the

induction

of

partial

EMT.

Such

events

could

account,

at

least

in

part,

for

the

carcinogenic

potential

of

endosulfan.

2. Materialsandmethods

2.1. Materials

ThehumanhepatocellularcarcinomacellsHepG2wereobtainedfromATCC (AmericanTypeCultureCollection,Manassas,VA).Dulbecco’smodifiedEagle’s medium (DMEM), fetal bovine serum (FBS), penicillin/streptomycinsolution, sodiumpyruvateandEagle’snon-essentialaminoacidswerefromBioWhittaker (CambrexCompany,Walkersville,USA).DMSO(dimethylsulfoxide)andchemicals werefromSigma–Aldrich(L’Isled’AbeauChesne,SaintQuentinFallavier,France). ProteinassaymaterialswerefromBio-Rad.Allfluorescencereagentswerefrom MolecularProbes(Eugene,OR).TheOCendosulfanwasfromChemService(West Chester,PA).Theantibodiesusedforwesternblottingandimmunodetection exper-imentswerefromCellSignaling,SantaCruzandEpitomics(Table1).Cellswere visualizedwithaNikonEclipseTE2000phasecontrastmicroscope.

2.2. Cellcultureanddrugtreatments

HepG2cellsweremaintainedinDMEMwith1%penicillin/streptomycin,1%non essentialaminoacids,sodiumpyruvate,and10%FBS,inhumidifiedatmosphereat 37◦Ccontaining95%O2and5%CO2.Afterwashingwithsterilephosphatebuffer

saline(PBS),cellsweredetachedbytrypsinization(trypsin/EDTA)andplatedata concentrationof0.5–1×106or0.4–0.5×105cells/mlin6-wellor24-wellplates,

respectively,dependingontheexperiment.Forallexperimentalconditions,FBS wasreducedto5%inDMEMmediumandcellsweretreatedfortheindicatedtime withendosulfanpreparedasdimethylsulfoxide(DMSO)stocksolution.Thefinal DMSOconcentrationwas0.25%(v/v).

2.3. Viabilitytest

Viablecells were determinedby measuring theconversion ofthe tetra-zoliumsaltMTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide, Sigma–Aldrich(St.Louis,MO)toformazan,aspreviouslydescribed(Fautreletal., 1991).Briefly,cellswereseededin96wellplatesandtreatedat50%confluency withaconcentrationof20␮Mendosulfanduring24,48and72h.Thecellswere thenincubatedwith0.5mg/mlMTTfor2hat37◦C.Thewater-insolubleformazan

crystalwasdissolvedbyadding100␮lDMSOtoeachwellandtheabsorbancewas determinedwithaspectrophotometerat550nm(MR7000,DynatechLaboratories, Inc.,USA).

(4)

2.4. Real-timecellularimpedance

ThexCELLigencesystemwasusedaccordingtothemanufacturers’instructions (RocheAppliedScience,Mannheim,Germany)andACEABiosciences(SanDiego, CA,USA).Briefly,1×104cellswereaddedtothe96-wellE-plates.Twenty-four

hourslater,cellsweretreatedwithdifferentconcentrationsofendosulfan(2,10, 20,50,100and200␮M)in5%FBSDMEMmedium.Real-timecellularimpedance wasmeasuredineachwell(cellindexvalues)andasignalwasobservedthroughthe integratedsoftware(RTCAAnalyzer).Eachcurveisrepresentativeofanexperiment performedintriplicate.Tocomparetheinfluenceoftheendosulfanonthecells,the normalizedcellindex(NCI)wasused,calculatedusingtheDMSOcontrolcondition.

2.5. Immunofluorescence

Cellswereseededonglassslidesin24-wellplates(4×105cells perwell)

andwereexposedtoendosulfanattheindicatedtimeandconcentration.For cytoskeletonandextra-cellularmatrixproteins(Rho-associatedkinase,vimentin andfibronectin)andtranscriptionfactors(Snail,SlugandHif-1␣),thecellswere fixedwith4%paraformaldehyde(PFA)andpermeabilizedin0.5%saponin.For struc-turalproteins(E-cadherinand␤-catenin),cellswerefixedin−20◦Cmethanoland

permeabilizedin0.25%triton.Inbothcases,slideswereincubatedwithantibodies (Table1)for1hatroomtemperature.AfterwashingwithPBS,goatanti-rabbitor anti-mouseIgGcoupledtoAlexaFluor®488or594(MolecularProbes,Eugen,OR)

wereaddedtoslides,beforeincubationinadarkroomfor1hatroom tempera-ture.Theactincytoskeletonwasobservedafterincubationwithphalloidincoupled withAlexaFluor488for5min,andthenucleusafterincubationwithDAPI1␮g/ml (2,6-diamidino-2-phenylindone)for10min.Slidesweremountedandsealedin ProLongGoldantifadereagent(Invitrogen).Observationswereperformedwithan invertedfluorescencemicroscope(Nikon)equippedwithaCDDcamera(ORCA-ER HamamatsuPhotonics).

2.6. Detectionoftheanoikisprocess

Thecellsweretreatedfor48or72hwith2,10,20,50and100␮Mofendosulfan. Then,theculturemediacontainingfloatingcellswerecollected,centrifuged,and pelletsreseededintonew6-wellplateswithDMEMsupplementedwith5%FBS.This protocolisdesignedtotesttheabilityofcellstoadhereintheabsenceofendosulfan. Eighteendayslater,theMTTviabilitytestwasperformedtoestimatethenumber ofnon-adheringcellsfromtheobservedpopulationofadheringcells.

Secondly,todeterminethelevelofanoikis,2×105cellswereculturedasa

sus-pensiononplatescoatedwith(poly(2-hydroxyethylmethacrylate)(poly-HEMA) (Sigma)fortheindicatedperiodsoftime.Apoptoticcellswerethenestimatedby enzymaticassayusingcaspase-3activityasamarkerofapoptosis.

2.7. Enzymaticassaysforcaspaseactivity

Caspase activity was assessed by measuring fluorophore (7-amido-4-trifluoromethylcoumarin (AFC)) release from caspase tetrapeptide substrate N-acetyl-Asp-Glu-Val-Asp (Ac-DEVD) for caspases-3-like activity. Briefly, cells grownin6-wellculturedisheswerescrapedintoice-coldhypotonicbuffer.Cells werethenlysedbybeingsubjectedtothreecyclesoffreezingandthawing.Protein concentrationsweredeterminedusingtheBCAProteinAssaykit(Pierce,Rockford, IL,USA)andequalamountsweremixedwithbufferB(312.5mMHEPES,pH7.5, 31.25%sucrose,0.3125%CHAPS,50␮Mofrelativesubstrateenzymes).The fluo-rometricassaydetectedtheshiftinAFCfluorescenceemissionfollowingcleavage fromtetrapeptide-AFC,asmeasuredinafluorometer(ex=390nm;em=530nm).

2.8. Westernblot

HepG2cellswerescrapedintohypotonicbuffer(20mMHEPES,pH7.5,10mM KCl,15mMMgCL2,0.25mMsucrose,1mMEDTA,1mMEGTA,1mMDTT,0.1mM

PMSF,10␮g/mlpepstatinA,10␮g/mlleupeptin,andthephosphataseinhibitory cocktailPhosphoSTOP,Roche).Theproteinconcentrationineachcelllysatewas quantifiedusingaBCAProteinAssayKit(Pierce),withbovineserumalbumin(BSA) usedasastandard.EqualproteinamountswereseparatedbySDS-polyacrylamide gelelectrophoresison10%gelsandweretransferredtoPVDFmembranes.The membraneswereimmunoblottedwithantibodies(Table1)for1hatroom tem-peratureorovernight at4◦C.Afterwashing,membraneswerethenincubated

withhorseradishperoxidase-conjugatedsecondaryantibodies(anti-mouseor anti-rabbitimmunoglobulinG;Promega,Madison,WI,USA)for1hatroomtemperature. Afterwashing,thesignalsweredetectedusingImmobilonWesternDetection Reagents(Millipore,Molsheim,France)andacquiredusingaCCDcamera (Chemi-Genius2,SynGene).

2.9. Reversetranscription-quantitativepolymerasechainreaction

TotalRNAwasisolatedusingacidphenolextraction.OnemicrogramoftotalRNA wasreversetranscribedusingakit(SuperScriptII;InvitrogenCorp.,Carlsbad, Cal-ifornia)followingthemanufacturer’sinstructions.QuantitativePCRanalysiswas

carriedoutwithLightCycler®480ProbesMaster(Roche),accordingtothe

man-ufacturer’sinstructions,togetherwithFAM-labeledhydrolysisprobesfromthe UniversalHumanProbeLibrarySet(Roche).Intron-spanningprimersweredesigned usingtheUniversalProbeLibraryAssayDesignCentersoftware.Calculationswere madeusinggapdhastheendogenouscontrolreferencegene.Folddifferencesingene expressionwerecalculatedusingtheLightCyclersoftware,takingintoaccountthe efficiencyofamplificationasdeterminedfromastandardcurveobtainedwiththe second-derivativemaximummethod.

2.10. Cellmigrationassay

At90%confluenceHepG2cellsweretrypsinedand3×106ofcellsperwell

wereaddedtoa6wellplate.After24hofincubation,theconfluenttissueformed wasscratched4timesperwellwithasterilepipettetip.Cellswerewashedtwice withPBSmediumbeforebeingtreatedwith0.25%DMSOor20␮Mendosulfanina DMEMmediumdepletedwith5%FBS.Imagesweretakenimmediately(0h)with aninvertedfluorescencemicroscope(Nikon)equippedwithaCDDcamera (ORCA-ERHamamatsuPhotonics)andNIS-ElementsAR2.30softwareat4×magnification. 24hlater,thecellmigrationprogresswasphotographedinthesameconditions andalldataweretreatedwiththeTScratchsoftwaretooldevelopedforautomated analysisofmonolayerwoundhealingassaysasdescribedbyGebäcketal.(2009).

2.11. Statisticalanalysis

Eachexperimentwasrepeatedatleastthreetimes.Datashownarean aver-age±standarddeviation(SD).Statisticalanalysisofinvitrostudieswasperformed usingaStudent’sttest.Levelsofprobabilityareindicatedas*P<0.05or**P<0.01.

3.

Results

3.1.

Morphological

changes

in

HepG2

cells

after

endosulfan

treatment

To

assess

the

effect

of

endosulfan

on

hepatoma

cells,

HepG2

cells

were

treated

with

increasing

concentrations

of

the

organochlorine

pesticide.

Viability

was

evaluated

by

MTT

dye

reduction

assay

after

24,

48

and

72

h

of

treatment

or

by

measuring

cellular

impedance

in

real-time.

As

shown

in

Fig.

1

,

endosulfan

decreased

HepG2

viabil-ity

in

a

dose-

and

time-dependent

manner.

Cytotoxicity

occurred

within

72

h

following

50

␮M

endosulfan

and

peaked

after

100

␮M

(IC

50

=

68.9

␮M

after

72

h

treatment).

Results

obtained

with

the

xCELLigence

system

do

not

allow

the

determination

of

IC50

values.

Indeed,

this

technology

is

a

non-invasive

cytotoxicity

assay

that

is

based

on

the

measurement

of

impedance

in

real

time

(cell

index

values).

This

parameter

reflects

the

cellular

status

with

regards

to

such

as

cell

proliferation,

vari-ations

in

cell

membrane

integrity,

cytotoxicity,

cell

adhesion

and

spreading,

and

cell

migration.

Fig.

1

B

depicts

the

dynamic

changes

in

cell

index

(CI)

values

of

cells

after

exposure

to

different

concen-trations

of

endosulfan.

At

100

and

200

␮M,

endosulfan

lead

to

a

slight

decline

in

the

cell

survival

rate

but

failed

to

induce

100%

mortality.

At

lower

concentrations,

a

significant

increase

in

the

slope

corresponding

to

the

evolution

of

cell

index

over

time

was

observed

(

Fig.

1

B

and

C).

This

phenomenon

could

be

explained

by

morphological

changes

correlating

with

defects

in

cell

adhesion

or

migration.

In

the

control

condition

(DMSO),

HepG2

cells

had

an

epithelial

cell-like

morphology

with

a

characteristic

“cobblestone”

appear-ance

and

organized

cortical

pattern

of

F-actin

at

cell-to-cell

junctions

(

Fig.

1

D).

By

contrast,

HepG2

cells

exposed

to

20

␮M

endosulfan

treatment

exhibited

more

poorly

defined

intercellular

borders

and

were

more

often

spherical

and

individualized

(arrows).

These

modifications

correlated

with

the

reduced

organization

of

F-actin

into

a

cortical

pattern

at

cell-to-cell

junctions

(

Fig.

1

D,

arrowhead).

These

results

suggested

that

endosulfan

is

toxic

at

high

concen-trations

(>50

␮M)

but

that

lower

doses

could

nevertheless

induce

phenotypic

modifications.

(5)

Fig.1.EndosulfanmodifiestheHepG2cellmorphology.(A)HepG2cellviabilitywasassessedbyMTTtestafter24h,48hand72htreatmentwithincreasingconcentrations ofendosulfan(from0.1␮Mto200␮M).MTTresultsarepresentedasapercentageofviabilityoverDMSOtreatmentandeachvalueisthemean±S.D.ofthreeseparate experiments(note:means±S.D.,*P<0.05and**P<0.001whencomparedtoDMSO).(BandC)Cellswereseededonto96-wellE-platesandtreatedfor24hwithendosulfan (from2,10,20,50,100and200␮M).Cellimpedancewasmeasuredinreal-timeandcellindexnormalizedagainsttheDMSOcondition.Resultsaremeans±S.D.for triplicatesofoneexperimentandarerepresentativeofthreeindependentexperiments.(D)48hafter20␮Mendosulfantreatment,cellmorphologywasexaminedundera lightmicroscope.F-actinwasvisualizedbyAlexaFluor488-conjugatedphalloidinstaining(green)andfluorescencemicroscopy.(Forinterpretationofthereferencestocolor inthisfigurelegend,thereaderisreferredtothewebversionofthearticle.)

3.2.

Endosulfan

sensitizes

HepG2

cells

to

anoikis

Based

upon

our

morphological

findings,

we

sought

to

evalu-ate

whether

endosulfan

affected

cell

death

processes.

We

observed

that

nuclei

from

individualized

cells

after

exposure

to

20

and

50

␮M

endosulfan

were

condensed

and

fragmented,

as

shown

by

DAPI

staining

(

Fig.

2

A).

Moreover,

when

these

floating

cells

were

replated

in

tissue

culture

dishes

after

48

h

or

72

h

in

suspension,

they

dis-played

the

ability

to

attach

and

proliferate

(

Fig.

2

B).

Indeed,

18

days

after

having

replated

the

cells,

MTT

test

showed

that

the

floating

cells

obtained

after

endosulfan

treatment

(<50

␮M)

had

grown

in

a

time-

and

dose-dependent

manner.

However,

at

high

concentra-tions

(50

and

100

␮M),

the

number

of

living

floating

cells

seemed

to

be

too

low,

as

demonstrated

by

the

low

viability

rate

obtained

after

replating

(

Fig.

2

C).

Hence,

these

results

suggest

that

the

process

leading

to

their

individualization

is

reversible.

We

firstly

inves-tigated

the

effect

of

endosulfan

on

capase-3

activity

levels

(as

a

marker

of

apoptosis)

in

both

the

adherent

and

the

floating

cells

after

48

h

endosulfan

treatment

(

Fig.

3

A).

The

caspase-3

activity

remained

stable

in

the

HepG2

cells

of

the

monolayer,

whereas

it

increased

significantly

in

the

floating

cells

at

concentrations

≥10

␮M

(400–450%

of

activation/DMSO).

Secondly,

to

investigate

whether

prevention

of

attachment

was

the

main

cause

of

cell

death

induced

by

endosulfan,

HepG2

cells

were

cultured

on

poly-HEMA

(+poly-HEMA),

which

prevented

cells

from

attaching.

The

cells

responded

with

a

significant

induction

of

caspase-3

activity

when

exposed

to

20

or

50

␮M

endosulfan

for

4

h

and

24

h,

but

not

with

DMSO

or

when

kept

under

adherent

conditions

(

−poly-HEMA).

However,

this

increased

caspase-3

activity

seemed

to

be

transient,

as

it

was

not

observed

after

48

h

treatment

(data

not

shown).

Moreover,

after

24

h

endosulfan

treatment

(20

␮M)

we

observed

an

induction

of

the

gene

encoding

X-linked

inhibitor

of

apoptosis

(XIAP),

the

endogenous

caspase-3

and

-7

inhibitor.

Thus,

endosulfan

initially

sensitized

the

HepG2

cells

to

anoikis

yet

per-mitted

the

restoration

of

intrinsic

anoikis

resistance

later

on.

Taken

together,

these

findings

demonstrate

that

endosulfan

may

induce

anoikis

in

HepG2

cells,

probably

via

disruption

of

cell

attachment.

3.3.

Endosulfan

induces

changes

in

the

cytoskeleton

and

ECM

associated

with

the

activation

of

FAK

signaling

and

potential

destabilization

of

adherent

junctions

We

therefore

investigated

whether

endosulfan

can

perturb

cel-lular

adhesion

by

studying

changes

in

the

expression

of

proteins

implicated

in

the

modification

of

the

actin

cytoskeleton

of

HepG2

cells

exposed

to

endosulfan.

We

examined

the

localization

and

expression

of

the

Rho-associated

kinase

(ROCK1),

a

well-known

effector

of

the

RhoGTPAses

implicated

in

lamelliopodia

and

stress

fiber

formation

during

cytoskeleton

remodeling

(

Schaller,

2010

).

(6)

Fig.2.Effectsofendosulfanonadherence,viabilityandnuclearcondensationofHepG2cells.(A)NuclearmorphologywasobservedbyDAPIstainingafter24hand48h ofDMSO(0.25%)orendosulfantreatments(20and50␮M).Arrowsindicateapoptoticnucleifromnon-adherentcellsafterendosulfantreatment.Theresultsshownare representativeofthreeseparateexperiments.(BandC)Floatingcellsobtainedafter48hor72hof20␮Mendosulfanexposurewerereplatedin6-wellplatesasdescribed inthematerialandmethodssection.18daysafterplating,picturesweretaken(B)andcellviabilitywasmeasuredbyMTTtest(C).

As

demonstrated

by

immunofluorescence

(

Fig.

4

A),

the

cellular

localization

of

ROCK1

was

modified

after

48

h

of

treatment

with

20

␮M

endosulfan.

Indeed,

in

the

control

condition

(DMSO),

ROCK1

was

mainly

colocalized

with

actin

leading

to

a

cortical

pattern

at

cell-to-cell

junctions,

whereas

in

the

presence

of

endosulfan,

this

protein

was

distributed

uniquely

throughout

the

cytoplasm.

We

also

examined

the

protein

levels

of

ROCK1

by

western

blot

analy-sis.

After

endosulfan

treatment,

the

expression

of

this

protein

was

up-regulated

in

a

time-dependent

manner

(

Fig.

4

B).

Focal

adhesion

kinase

(FAK)

signaling

was

also

implicated

in

the

control

of

the

actin

cytoskeleton

organization.

Indeed,

FAK

medi-ates

cells

motility

and

adhesion

turnover

through

regulation

of

the

RhoGTPases

and

ROCK

activity

(

Riento

and

Ridley,

2003

).

We

found

that

endosulfan

increased

the

phosphorylation

of

FAK

at

Tyr

925

in

a

time-dependent

manner

as

soon

as

8

h

of

endosulfan

treatment

(

Fig.

4

C).

The

phosphorylation

of

the

Tyr-925

residue

leads

to

cell

migration

and

cell

protrusion

(

Deramaudt

et

al.,

2011

).

Extracellular

matrix

(ECM)

remodeling

is

a

major

pheno-typic

modification

observed

during

cell

motility

and

migration.

Fibronectin,

a

high-molecular

weight

glycoprotein,

serves

as

a

scaf-fold

for

the

fibrillar

ECM.

We

therefore

analyzed

fibronectin

levels

by

western

blotting

and

immunofluorescence

studies.

Immunoflu-orescence

analysis

showed

an

important

increase

of

fibronectin

in

the

cytoplasm

of

HepG2

cells

after

48

h

of

endosulfan

treatment

(

Fig.

4

D).

These

findings

are

consistent

with

the

up-regulation

of

fibronectin

detected

by

western

blot

(

Fig.

4

E).

It

is

worth

noting

that

the

protein

level

of

fibronectin

was

increased

after

30

min

of

endosulfan

treatment

and

peaked

after

about

60

min.

Moreover,

fibronectin

was

deposited

as

fibrils

(

Fig.

4

F)

in

the

extracellular

compartment,

48

h

after

endosulfan

exposure.

Interestingly,

endo-sulfan

did

not

increase

the

gene

expression

of

Itga5,

corresponding

to

the

␣5

chain

of

the

fibronectin

receptor

integrin

␣5␤1

(data

not

shown).

This

integrin

is

the

main

receptor

of

fibronectin

allowing

cells

to

migrate.

Together,

these

results

show

that

endosulfan

induced

cytoskele-ton

remodeling

and

a

modification

of

the

ECM

composition.

3.4.

Loss

of

adherent

junctions

through

E-cadherin

repression:

possible

involvement

of

the

repressors

Snail

and

Slug

To

further

confirm

the

effect

of

endosulfan

on

cell

adhesion,

we

investigated

E-cadherin

gene

and

protein

expression

levels

in

HepG2

cells

exposed

to

endosulfan.

Endosulfan

significantly

decreased

the

protein

levels

of

E-cadherin

after

24

h

and

72

h

of

endosulfan

treatment

(

Fig.

5

A).

Moreover,

E-cadherin

mRNA

expression

was

decreased

by

approximately

50%

after

48

h

endo-sulfan

treatment

(

Fig.

5

B).

These

data

suggested

that

E-cadherin

expression

is

repressed

in

HepG2

cells

at

the

transcriptional

level

after

endosulfan

exposure.

Hence,

we

sought

to

determine

the

expression

and

the

localization

of

Snail

and

Slug,

two

direct

(7)

Fig.3. Inductionofanoikisbyendosulfantreatment.(AandB)HepG2wereexposedtoincreasedconcentrationsofendosulfan(from2to50␮M)for48h.Caspase-3activities wereassayedasdescribedintheexperimentalproceduresineitheradherentcellsofthecellmonolayerorinfloatingcells(non-adherentcells)(A).(B)HepG2cellswere culturedovernighteitheradherent(−poly-HEMA)orsuspendedonpoly-HEMA-coatedplates(+poly-HEMA)andreceivedendosulfan(20or50␮M)for4,24or48hbefore measuringcaspase-3activity.(AandB)ResultsareexpressedasapercentageofthevalueobtainedforDMSO-treatedcells,setat100%.Dataaremeans±SDforfourseparate experiments.Caspase-3activitieswereassayedasdescribedintheexperimentalprocedures.(C)XIAPmRNAlevelwasassessedbyreal-timeRT-PCRafter4,24and48hof 20␮Mendosulfantreatment.RelativemRNAexpressionlevels(normalizedwithrespecttogapdh)weredeterminedandmRNAlevelsinDMSO-treatedcellsweresetto1. Errorbarsindicatethemeans±SEMoftriplicatedeterminationsfromthreeindependentexperiments.Note:*P>05and**P>0.01.

repressors

of

E-cadherin

transcription.

We

found

that

endosulfan

increased

Slug

and

Snail

mRNA

after

24

h

and

48

h,

respectively

(

Fig.

5

C).

Concomitantly,

we

observed

a

nuclear

translocation

of

these

two

factors

into

the

nucleus

of

HepG2

cells

treated

with

endosulfan

(

Fig.

5

D).

Since

a

decrease

in

levels

of

E-cadherin

is

a

widely

accepted

characteristic

associated

with

EMT,

we

wondered

whether

endo-sulfan

could

activate

this

process.

3.5.

Endosulfan

induces

stabilization

and

nuclear

translocation

of

ˇ-catenin

During

EMT,

loss

of

E-cadherin-mediated

cell–cell

adhesion

destabilizes

the

␤-catenin

interaction

with

actin

cytoskeleton

and

can

promote

its

activation.

Accordingly,

we

sought

to

evaluate

whether

endosulfan

could

act

on

the

␤-catenin

signaling

pathway

(Clevers,

2006).

After

endosulfan

treatment,

the

protein

level

of

␤-catenin

increased

in

a

time-dependent

manner

(

Fig.

6

A)

without

significant

modulation

of

its

gene

expression

(

Fig.

6

B).

This

protein

stabiliza-tion

occurred

together

with

the

nuclear

translocation

of

␤-catenin

after

48

h

endosulfan

treatment,

as

shown

by

immunofluorescence

imaging

in

Fig.

6

C.

Once

in

the

nucleus,

␤-catenin

binds

with

transcription

factors,

such

as

LEF/TCF,

to

activate

different

targets

genes,

such

as

LEF1,

TCF-1,

MMP-7

or

CCND1

(cyclin-D1)

(

Roose

and

Clevers,

1999;

Wielenga

et

al.,

1999;

Brabletz

et

al.,

1999;

Tetsu

and

McCormick,

1999

).

Endosulfan

increased

in

a

time-dependent

manner

the

protein

level

of

LEF1/TCF-1

when

compared

to

the

DMSO

condition

(

Fig.

6

D).

Moreover,

we

found

that

endosulfan

up-regulated

both

MMP-7

and

CCND1

at

the

mRNA

level

after

48

h

treatment.

These

results

suggest

that

endosulfan

activates

the

WNT/

␤-catenin

signaling

pathway.

3.6.

Endosulfan

induces

gain

of

the

mesenchymal

markers

S100a4

and

vimentin

but

does

not

confer

the

ability

to

migrate

EMT

is

characterized

by

loss

of

epithelial

and

gain

of

mes-enchymal

markers.

Consequently,

we

investigated

the

effect

of

endosulfan

on

the

expression

of

S100a4,

a

marker

of

hepatocytes

undergoing

EMT,

and

vimentin,

an

intermediate

filament

protein

normally

found

in

cells

of

mesenchymal

origin.

As

demonstrated

by

real-time

PCR,

endosulfan

increased

tran-siently

S100a4

gene

expression,

as

soon

as

4

h

and

peaked

after

24

h

endosulfan

treatment

(

Fig.

7

A).

Moreover,

we

observed

increased

amounts

of

vimentin

in

the

cytoplasm

of

HepG2

cells

exposed

to

endosulfan

(

Fig.

7

B).

These

results

would

suggest

that

endosulfan

leads

to

a

con-version

of

HepG2

cells

to

a

mesenchymal

phenotype

with

the

capacity

to

migrate.

Yet,

wound-healing

assays

revealed

no

(8)

Fig.4. EndosulfanperturbsthecytoskeletoninHepG2cellsandtheextracellularmatrix.(A,DandF)CellsweregrownoncoverslipsandtreatedeitherwithDMSO(0.25%) or20␮Mendosulfan.48hlater,cellswerefixedandprocessedforindirectimmunofluorescenceforthedetectionofactin(AandD,green),ROCK1(A,red)andfibronectin (DandF,red).Colocalizationwasindicatedbytheyellowcolorinthemergedimage.Representativeofthreeseparateexperiments.(B,CandE)HepG2cellsweretreated with20␮Mendosulfan.Attheindicatedtime,cellswerelysedandproteinlevelsoffibronectine(E),phospho-Fak,Fak(C)andRock1(B)analyzedbywesternblotting.As acontrol,thesamemembraneswerealsoprobedwithanantibodydirectedagainstGapdh.Thewesternresultsarerepresentativeofthreeindependentrepeatsforeach experiment.(Forinterpretationofthereferencestocolorinthisfigurelegend,thereaderisreferredtothewebversionofthearticle.)

significant

increase

in

wound

closure

after

24

h

or

48

h

endosulfan

treatment

when

compared

to

the

DMSO

condition

(

Fig.

7

C).

Thus

although

endosulfan

is

able

to

induce

major

changes

with

regards

to

the

expression

of

epithelial

characteristics,

it

does

not

confer

migration

properties

and

therefore

appears

to

induce

a

par-tial

or

incomplete

EMT

in

HepG2

cells.

4.

Discussion

In

this

study,

we

have

shown

that

endosulfan

transiently

sen-sitizes

HepG2

cells

to

anoikis,

disrupts

the

epithelial

phenotype

of

HepG2

and

induces

a

partial

EMT

process.

Anoikis

is

defined

as

cell

death

induced

by

inappropriate

or

loss

of

cell

adhesion

(

Frisch

and

Francis,

1994;

Meredith

et

al.,

1993

).

HepG2

carcinoma

cells

are

resistant

to

anoikis

but

after

endosul-fan

treatment

we

observed

an

induction

of

caspase-3

activity

in

poly-HEMA

culture

within

the

first

24

h.

After

that,

cells

recov-ered

their

resistance

to

anoikis

as

shown

by

increased

XIAP

mRNA

levels.

This

endogenous

caspase

inhibitor

protein

has

been

shown

to

contribute

to

the

anoikis

resistance

of

various

carcinoma

cells

(

Berezovskaya

et

al.,

2005;

Liu

et

al.,

2006

).

Hence,

overexpression

of

XIAP

in

cancer

cells

is

linked

to

their

increased

resistance

to

apo-ptosis

and

the

expression

level

reflects

apoptosis

sensitivity

(

Shi

et

al.,

2008

).

Acquisition

of

anoikis

resistance

is

a

likely

prerequisite

for

tumor

cells

to

successfully

metastasize

to

distant

sites

(

Liotta

and

Kohn,

2004

).

The

loss

of

cellular

contact

with

the

basement

membrane

constitutes

the

starting

point

of

the

onset

of

anoikis.

This

cell–ECM

interaction

is

monitored

by

integrins

that

bind

to

diverse

ECM

molecules

and

respond

by

triggering

an

intracellular

cascade

via

SRC

and

FAK

kinases.

We

found

that

endosulfan

increases

the

phosphorylation

of

FAK

at

the

Tyr

925

residue,

an

event

likely

to

contribute

toward

the

activation

of

downstream

signaling

events

including

the

Rho

GTPase

pathway.

This

sustained

phosphorylation

of

FAK

after

endosulfan

exposure

correlated

with

the

overexpres-sion

and

relocalization

of

the

cytoskeletal

RhoGTPase

proteins

(data

not

shown)

and

their

main

effector

ROCK1.

This

cascade

has

been

linked

to

cancer

cell

migration/invasion

during

metastasis

via

the

control

of

cytoskeletal

remodeling

(

Yamazaki

et

al.,

2005

).

FAK

is

a

major

protein

of

the

focal

adhesion

complex

that

integrates

signals

from

growth

factors

and

integrins

to

control

ECM

inter-actions,

migration

and

invasion

(

Mitra

et

al.,

2005

).

This

family

of

kinases

was

linked

to

tumor

invasiveness

and

their

expres-sion

and/or

activation

is

frequently

associated

with

metastatic

tumors

(

Gabarra-Niecko

et

al.,

2003;

McLean

et

al.,

2005

).

Notably,

in

parallel

to

FAK

phosphorylation,

we

found

an

up-regulation

of

fibronectin,

a

major

protein

that

serves

as

a

scaffold

for

the

fibrillar

ECM.

All

these

events

occurred

in

parallel

to

a

rearrange-ment

of

the

F-actin

cytoskeleton

with

formation

of

stress

fibers

in

(9)

Fig.5.EndosulfanrepressesE-cadherinexpression.(AandC)E-cadherin(CDH1),SnailandSlugmRNAlevelswereassessedbyreal-timeRT-PCRafter4,24and48hof20␮M endosulfantreatment.RelativemRNAexpressionlevels(normalizedwithrespecttogapdh)weredeterminedandmRNAlevelsinDMSO-treatedcellsweresetto1.Error barsindicatethemeans±SEMoftriplicatedeterminationsfromthreeindependentexperiments.(B)Attheindicatedtime,cellswerelysedandE-cadherinproteinlevels wereassessedbywesternblotting.(B)BandintensitieswereassessedbydensitometryafterimageacquisitionwithaCCDcameraandtheresultsarepresentedastheratioof Gapdh-normalizedresultsfortreatedcellstothoseforDMSO-treatedcells(means±SDforthreeexperiments).(D)After48hexposure,thecellswerefixedandprocessedfor indirectimmunofluorescenceanalysisforthedetectionofslugorsnail(red)andvisualizationofnuclei(DAPI,blue).Theresultsshownarerepresentativeofthreeseparate experiments.*P<0.05and**P<0.01.(Forinterpretationofthereferencestocolorinthisfigurelegend,thereaderisreferredtothewebversionofthearticle.)

endosulfan-treated

cells.

This

phenomenon

has

aleady

been

described

during

cellular

stress

and

cell

migration

(

Pollard

and

Borisy,

2003

).

Thus,

endosulfan

appears

to

structurally

reorga-nize

the

cytoskeleton

and

to

modify

the

ECM

composition.

These

changes

were

accompanied

by

phenotypical

modifications

such

as

those

observed

during

cell

dedifferentiation

(

van

Zijl

et

al.,

2009a,b

).

The

EMT

process

has

been

shown

to

tightly

correlate

with

anoikis

resistance

(

Klymkowsky

and

Savagner,

2009;

Smit

et

al.,

2009

).

Genes

that

drive

EMT

(such

as

Snail,

Slug,

ZEB1/2,

and

Twist)

frequently

down-regulate

E-cadherin

and

confer

anoikis

resistance.

Indeed,

it

has

been

demonstrated

that

the

loss

of

E-cadherin

is

a

major

landmark

in

the

progression

of

EMT,

and

confers

to

epithelial

cells

a

resistance

to

anoikis

(

Fouquet

et

al.,

2004;

Kumar

et

al.,

2011

).

During

EMT,

loss

of

anchorage

is

a

critical

event

required

for

cell

migration

(Yilmaz

and

Christofori,

2010).

We

observed

a

repression

of

E-cadherin

from

24

h

of

endosulfan

treatment,

coinciding

with

the

time

at

which

the

basal

anoikis

resistance

status

was

observed.

Consistently

with

these

results,

we

found

that

endosulfan

could

both

overexpress

and

activate

the

E-cadherin

repressors

Snail1

and

Slug,

two

important

factors

for

EMT

induction

(

Polyak

and

Weinberg,

2009

).

The

Snail

family

members

are

most

widely

recognized

as

suppressors

of

E-cadherin

expression

and

regulate

other

aspects

of

the

EMT

phenotype,

such

as

increased

expression

of

fibronectin

and

protection

from

cell

death

(

Zeisberg

and

Neilson,

2009

).

The

disruption

of

E-cadherin-mediated

adhesion

is

thought

to

be

a

key

step

in

the

progression

of

hepatocarcinoma

(

Behrens,

1993;

Takeichi,

1993;

Christofori

and

Semb,

1999

).

Indeed,

E-cadherin

downregulation

in

HCC

is

associated

with

increased

tumor

size,

low

levels

of

histological

dif-ferentiation,

invasion

recurrence,

metastasis

and

poor

prognosis

(

Kozyraki

et

al.,

1996;

Yang

and

Weinberg,

2008

).

We

also

observed

a

stabilization

of

the

␤-catenin

protein

and

its

translocation

to

the

nucleus

in

HepG2

cells

treated

with

endosulfan.

E-cadherin

repression

led

to

its

disappearance

from

the

intercellu-lar

junctions

and

to

the

release

of

␤-catenin

into

the

cytoplasm.

The

membrane-unbound

␤-catenin

is

usually

phosphorylated

by

a

complex

responsible

for

its

degradation

through

the

ubiquitin-proteasome

system

(

Cowin

et

al.,

2005;

Peifer

and

Polakis,

2000

).

Docking

of

the

Wnt

ligand

to

its

Frizzled

(Fz)

receptor

triggers

activation

of

the

canonical

Wnt

pathway

and

allows

␤-catenin

to

accumulate

in

the

cytoplasm

and

translocate

to

the

nucleus.

There

it

functions

as

a

cofactor

for

members

of

the

Tcf/Lef

fam-ily

of

transcription

factors,

which

further

activate

the

transcription

Figure

Fig. 1. Endosulfan modifies the HepG2 cell morphology. (A) HepG2 cell viability was assessed by MTT test after 24 h, 48 h and 72 h treatment with increasing concentrations of endosulfan (from 0.1 ␮M to 200 ␮M)
Fig. 2. Effects of endosulfan on adherence, viability and nuclear condensation of HepG2 cells
Fig. 3. Induction of anoikis by endosulfan treatment. (A and B) HepG2 were exposed to increased concentrations of endosulfan (from 2 to 50 ␮M) for 48 h
Fig. 4. Endosulfan perturbs the cytoskeleton in HepG2 cells and the extracellular matrix
+4

Références

Documents relatifs

Intravenous drug addicts are a population at risk for infectious complications such as bacteremia, funge- mia, endocarditis, pneumonia, arthritis, osteomye- litis,

To prepare the cycles of voltage pulse pairs used for pump-probe experiments connect an arbitrary function generator with at least two channels to the tip (Figure 2). The

This account explains the presence of object agreement in the perfective in complete agreement reversal languages: the number probe on T remains free (after the person probe agrees

Using cell biology approaches, we show that recProt01230 is able to adhere to bovine host cells and interacts with proteins from the cell lysate and the &#34;membranes/

In addition, after four days exposure at 1.75 µg.L -1 of endosulfan, an increase of erythrocyte DNA breaks quantity was observed.. The results suggest

Plasma levels of endogenous melatonin were measured by liquid chromatography-tandem mass spectrometry in PH patients (n = 64, 43 pulmonary arterial hypertension (PAH) and 21

A list of publications issued by the Division of Building Research can be obtained on application to the Publications Section, Division of Building Research,

In this study, different marine vegetation was sampled in three different geographical areas ( Fig. 1 ): three species of macroalgae (Fucus vesiculosus, Porphyra sp. and Ulva